46 results on '"Roovers RC"'
Search Results
2. High-affinity recombinant phage antibodies to the pan-carcinoma marker epithelial glycoprotein-2 for tumour targeting
- Author
-
Roovers, RC, primary, Henderikx, P, additional, Helfrich, W, additional, van der Linden, E, additional, Reurs, A, additional, de Bruïne, AP, additional, Arends, JW, additional, de Leij, L, additional, and Hoogenboom, HR, additional
- Published
- 1998
- Full Text
- View/download PDF
3. Leveraging Vγ9Vδ2 T cells against prostate cancer through a VHH-based PSMA-Vδ2 bispecific T cell engager.
- Author
-
King LA, Veth M, Iglesias-Guimarais V, Blijdorp I, Kloosterman J, Vis AN, Roovers RC, Hulsik DL, Riedl T, Adang AEP, Parren PWHI, van Helden PM, de Gruijl TD, and van der Vliet HJ
- Abstract
Vγ9Vδ2 T cells constitute a homogeneous effector T cell population that lyses tumors of different origin, including the prostate. We generated a bispecific T cell engager (bsTCE) to direct Vγ9Vδ2 T cells to PSMA
+ prostate cancer (PCa) cells. The PSMA-Vδ2 bsTCE triggered healthy donor and PCa patient-derived Vγ9Vδ2 T cells to lyse PSMA+ PCa cell lines and patient-derived tumor cells while sparing normal prostate cells and enhanced Vγ9Vδ2 T cell antigen cross-presentation to CD8+ T cells. Vγ9Vδ2 T cell expressed NKG2D and DNAM-1 contributed to Vγ9Vδ2 T cell activation and tumor lysis at low PSMA-Vδ2 bsTCE concentrations. In vivo models confirmed the antitumor efficacy of the bsTCE and demonstrated a half-life of 6-7 days. Tissue-cross reactivity analysis was in line with known tissue distribution of PSMA and Vγ9Vδ2 T cells. Together these data show the PSMA-Vδ2 bsTCE to represent a promising anti-tumor strategy and supports its ongoing evaluation in a phase 1/2a clinical trial in therapy refractory metastatic castration-resistant PCa., Competing Interests: L.A.K., M.V.,and J.K. are/were funded by Lava Therapeutics NV. T.R., R.C.R., A.E.P.A., P.W.H.I.P., T.D.d.G.,and H.J.v.d.V. own Lava Therapeutics NV shares. V.I.-G., I.B., R.C.R., D.L.H., T.R., A.E.P.A., P.W.H.I.P., and H.J.v.d.V. are/were employees of Lava Therapeutics NV. T.D.d.G.is scientific advisor to Lava Therapeutics NV. Patent registration related to this work: Antibodies that bind PSMA and gamma-delta T cell receptors, Lava Therapeutics NV, WO2022008646A1, 2022-01-13., (© 2024 The Authors.)- Published
- 2024
- Full Text
- View/download PDF
4. Structural insights into the role and targeting of EGFRvIII.
- Author
-
Bagchi A, Stayrook SE, Xenaki KT, Starbird CA, Doulkeridou S, El Khoulati R, Roovers RC, Schmitz KR, van Bergen En Henegouwen PMP, and Ferguson KM
- Subjects
- Humans, Crystallography, X-Ray, Models, Molecular, Glioblastoma metabolism, Protein Domains, Binding Sites, ErbB Receptors chemistry, ErbB Receptors metabolism, ErbB Receptors genetics, Single-Domain Antibodies chemistry, Single-Domain Antibodies metabolism, Protein Binding
- Abstract
The epidermal growth factor receptor (EGFR) is a well-known oncogenic driver in lung and other cancers. In glioblastoma multiforme (GBM), the EGFR deletion variant III (EGFRvIII) is frequently found alongside EGFR amplification. Agents targeting the EGFR axis have shown limited clinical benefits in GBM and the role of EGFRvIII in GBM is poorly understood. To shed light on the role of EGFRvIII and its potential as a therapeutic target, we determined X-ray crystal structures of a monomeric EGFRvIII extracellular region (ECR). The EGFRvIII ECR resembles the unliganded conformation of EGFR, including the orientation of the C-terminal region of domain II. Domain II is mostly disordered, but the ECR structure is compact. We selected a nanobody with preferential binding to EGFRvIII relative to EGFR and structurally defined an epitope on domain IV that is occluded in the unliganded intact EGFR. These findings suggest new avenues for EGFRvIII targeting in GBM., Competing Interests: Declaration of interests R.e.K. is an employee at TerThera and R.C.R. is an employee of Lava Therapeutics., (Copyright © 2024 Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
5. A Bispecific γδ T-cell Engager Targeting EGFR Activates a Potent Vγ9Vδ2 T cell-Mediated Immune Response against EGFR-Expressing Tumors.
- Author
-
King LA, Toffoli EC, Veth M, Iglesias-Guimarais V, Slot MC, Amsen D, van de Ven R, Derks S, Fransen MF, Tuynman JB, Riedl T, Roovers RC, Adang AEP, Ruben JM, Parren PWHI, de Gruijl TD, and van der Vliet HJ
- Subjects
- Humans, Mice, Animals, Leukocytes, Mononuclear, Receptors, Antigen, T-Cell, gamma-delta, Immunity, ErbB Receptors, Lymphocyte Activation, Neoplasms drug therapy, Antibodies, Bispecific pharmacology, Antibodies, Bispecific therapeutic use
- Abstract
Vγ9Vδ2 T cells are effector cells with proven antitumor efficacy against a broad range of cancers. This study aimed to assess the antitumor activity and safety of a bispecific antibody directing Vγ9Vδ2 T cells to EGFR-expressing tumors. An EGFR-Vδ2 bispecific T-cell engager (bsTCE) was generated, and its capacity to activate Vγ9Vδ2 T cells and trigger antitumor activity was tested in multiple in vitro, in vivo, and ex vivo models. Studies to explore safety were conducted using cross-reactive surrogate engagers in nonhuman primates (NHP). We found that Vγ9Vδ2 T cells from peripheral blood and tumor specimens of patients with EGFR+ cancers had a distinct immune checkpoint expression profile characterized by low levels of PD-1, LAG-3, and TIM-3. Vγ9Vδ2 T cells could be activated by EGFR-Vδ2 bsTCEs to mediate lysis of various EGFR+ patient-derived tumor samples, and substantial tumor growth inhibition and improved survival were observed in in vivo xenograft mouse models using peripheral blood mononuclear cells (PBMC) as effector cells. EGFR-Vδ2 bsTCEs exerted preferential activity toward EGFR+ tumor cells and induced downstream activation of CD4+ and CD8+ T cells and natural killer (NK) cells without concomitant activation of suppressive regulatory T cells observed with EGFR-CD3 bsTCEs. Administration of fully cross-reactive and half-life extended surrogate engagers to NHPs did not trigger signals in the safety parameters that were assessed. Considering the effector and immune-activating properties of Vγ9Vδ2 T cells, the preclinical efficacy data and acceptable safety profile reported here provide a solid basis for testing EGFR-Vδ2 bsTCEs in patients with EGFR+ malignancies., (©2023 American Association for Cancer Research.)
- Published
- 2023
- Full Text
- View/download PDF
6. A bispecific T cell engager recruits both type 1 NKT and Vγ9Vδ2-T cells for the treatment of CD1d-expressing hematological malignancies.
- Author
-
Lameris R, Ruben JM, Iglesias-Guimarais V, de Jong M, Veth M, van de Bovenkamp FS, de Weerdt I, Kater AP, Zweegman S, Horbach S, Riedl T, Winograd B, Roovers RC, Adang AEP, de Gruijl TD, Parren PWHI, and van der Vliet HJ
- Subjects
- Mice, Animals, T-Lymphocytes, Regulatory pathology, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Hematologic Neoplasms therapy, Leukemia, Myeloid, Acute
- Abstract
Bispecific T cell engagers (bsTCEs) hold great promise for cancer treatment but face challenges due to the induction of cytokine release syndrome (CRS), on-target off-tumor toxicity, and the engagement of immunosuppressive regulatory T cells that limit efficacy. The development of Vγ9Vδ2-T cell engagers may overcome these challenges by combining high therapeutic efficacy with limited toxicity. By linking a CD1d-specific single-domain antibody (VHH) to a Vδ2-TCR-specific VHH, we create a bsTCE with trispecific properties, which engages not only Vγ9Vδ2-T cells but also type 1 NKT cells to CD1d
+ tumors and triggers robust proinflammatory cytokine production, effector cell expansion, and target cell lysis in vitro. We show that CD1d is expressed by the majority of patient MM, (myelo)monocytic AML, and CLL cells and that the bsTCE triggers type 1 NKT and Vγ9Vδ2-T cell-mediated antitumor activity against these patient tumor cells and improves survival in in vivo AML, MM, and T-ALL mouse models. Evaluation of a surrogate CD1d-γδ bsTCE in NHPs shows Vγ9Vδ2-T cell engagement and excellent tolerability. Based on these results, CD1d-Vδ2 bsTCE (LAVA-051) is now evaluated in a phase 1/2a study in patients with therapy refractory CLL, MM, or AML., Competing Interests: Declaration of interests J.M.R., V.I.G., F.S.v.d.B., B.W., R.C.R., A.E.P.A., T.R., P.W.H.I.P., and H.J.v.d.V. are employees of LAVA Therapeutics, a company that develops bispecific gamma-delta T cell engagers, and own LAVA Therapeutics shares and/or stock options. R.L., T.D.d.G., P.W.H.I.P., and H.J.v.d.V. are named inventors on international patent application WO2020/060,405 ("Dual acting CD1d immunoglobulin"), which partially relates to the work described in this paper. T.D.d.G. is a consultant for and a shareholder of LAVA Therapeutics. A.P.K. is a consultant for LAVA Therapeutics. R.L., M.J., M.V., and I.d.W. were funded by a LAVA Therapeutics grant to Amsterdam UMC., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
7. Directing HIV-1 for degradation by non-target cells, using bi-specific single-chain llama antibodies.
- Author
-
Stam JC, de Maat S, de Jong D, Arens M, van Lint F, Gharu L, van Roosmalen MH, Roovers RC, Strokappe NM, Wagner R, Kliche A, de Haard HJ, van Bergen En Henegouwen PM, Nijhuis M, and Verrips CT
- Subjects
- Animals, Antibodies, Neutralizing, ErbB Receptors, HIV Antibodies, Humans, Camelids, New World, HIV Infections, HIV Seropositivity, HIV-1, Single-Chain Antibodies
- Abstract
While vaccination against HIV-1 has been so far unsuccessful, recently broadly neutralizing antibodies (bNAbs) against HIV-1 envelope glycoprotein were shown to induce long-term suppression in the absence of antiretroviral therapy in patients with antibody-sensitive viral reservoirs. The requirement of neutralizing antibodies indicates that the antibody mediated removal (clearance) of HIV-1 in itself is not efficient enough in these immune compromised patients. Here we present a novel, alternative approach that is independent of a functional immune system to clear HIV-1, by capturing the virus and redirecting it to non-target cells where it is internalized and degraded. We use bispecific antibodies with domains derived from small single chain Llama antibodies (VHHs). These bind with one domain to HIV-1 envelope proteins and with the other domain direct the virus to cells expressing epidermal growth factor receptor (EGFR), a receptor that is ubiquitously expressed in the body. We show that HIV envelope proteins, virus-like particles and HIV-1 viruses (representing HIV-1 subtypes A, B and C) are efficiently recruited to EGFR, internalized and degraded in the lysosomal pathway at low nM concentrations of bispecific VHHs. This directed degradation in non-target cells may provide a clearance platform for the removal of viruses and other unwanted agents from the circulation, including toxins, and may thus provide a novel method for curing., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
8. Functional patient-derived organoid screenings identify MCLA-158 as a therapeutic EGFR × LGR5 bispecific antibody with efficacy in epithelial tumors.
- Author
-
Herpers B, Eppink B, James MI, Cortina C, Cañellas-Socias A, Boj SF, Hernando-Momblona X, Glodzik D, Roovers RC, van de Wetering M, Bartelink-Clements C, Zondag-van der Zande V, Mateos JG, Yan K, Salinaro L, Basmeleh A, Fatrai S, Maussang D, Lammerts van Bueren JJ, Chicote I, Serna G, Cabellos L, Ramírez L, Nuciforo P, Salazar R, Santos C, Villanueva A, Stephan-Otto Attolini C, Sancho E, Palmer HG, Tabernero J, Stratton MR, de Kruif J, Logtenberg T, Clevers H, Price LS, Vries RGJ, Batlle E, and Throsby M
- Subjects
- ErbB Receptors metabolism, Humans, Imidazoles, Neoplastic Stem Cells metabolism, Organoids, Pyrazines, Receptors, G-Protein-Coupled metabolism, Antibodies, Bispecific pharmacology, Neoplasms, Glandular and Epithelial metabolism
- Abstract
Patient-derived organoids (PDOs) recapitulate tumor architecture, contain cancer stem cells and have predictive value supporting personalized medicine. Here we describe a large-scale functional screen of dual-targeting bispecific antibodies (bAbs) on a heterogeneous colorectal cancer PDO biobank and paired healthy colonic mucosa samples. More than 500 therapeutic bAbs generated against Wingless-related integration site (WNT) and receptor tyrosine kinase (RTK) targets were functionally evaluated by high-content imaging to capture the complexity of PDO responses. Our drug discovery strategy resulted in the generation of MCLA-158, a bAb that specifically triggers epidermal growth factor receptor degradation in leucine-rich repeat-containing G-protein-coupled receptor 5-positive (LGR5+) cancer stem cells but shows minimal toxicity toward healthy LGR5+ colon stem cells. MCLA-158 exhibits therapeutic properties such as growth inhibition of KRAS-mutant colorectal cancers, blockade of metastasis initiation and suppression of tumor outgrowth in preclinical models for several epithelial cancer types., (© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.)
- Published
- 2022
- Full Text
- View/download PDF
9. A Bispecific Antibody Antagonizes Prosurvival CD40 Signaling and Promotes Vγ9Vδ2 T cell-Mediated Antitumor Responses in Human B-cell Malignancies.
- Author
-
de Weerdt I, Lameris R, Scheffer GL, Vree J, de Boer R, Stam AG, van de Ven R, Levin MD, Pals ST, Roovers RC, Parren PWHI, de Gruijl TD, Kater AP, and van der Vliet HJ
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Antibodies, Bispecific immunology, Cell Line, Tumor, Female, HEK293 Cells, Humans, Leukemia, Lymphocytic, Chronic, B-Cell pathology, Lymphocyte Activation drug effects, Male, Mice, Mice, Inbred NOD, Middle Aged, Signal Transduction drug effects, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antibodies, Bispecific pharmacology, CD40 Antigens immunology, Leukemia, Lymphocytic, Chronic, B-Cell drug therapy, Leukemia, Lymphocytic, Chronic, B-Cell immunology
- Abstract
Novel T cell-based therapies for the treatment of B-cell malignancies, such as chronic lymphocytic leukemia (CLL) and multiple myeloma (MM), are thought to have strong potential. Progress, however, has been hampered by low efficacy and high toxicity. Tumor targeting by Vγ9Vδ2 T cells, a conserved T-cell subset with potent intrinsic antitumor properties, mediated by a bispecific antibody represents a novel approach promising high efficacy with limited toxicity. Here, we describe the generation of a bispecific Vγ9Vδ2 T-cell engager directed against CD40, which, due to its overexpression and biological footprint in malignant B cells, represents an attractive target. The CD40-targeting moiety of the bispecific antibody was selected because it can prevent CD40L-induced prosurvival signaling and reduce CD40-mediated resistance of CLL cells to venetoclax. Selective activation of Vγ9Vδ2 T cells in the presence of CD40
+ tumor cells induced potent Vγ9Vδ2 T-cell degranulation, cytotoxicity against CLL and MM cells in vitro , and in vivo control of MM in a xenograft model. The CD40-bispecific γδ T-cell engager demonstrated lysis of leukemic cells by autologous Vγ9Vδ2 T cells present in patient-derived samples. Taken together, our CD40 bispecific γδ T-cell engager increased the sensitivity of leukemic cells to apoptosis and induced a potent Vγ9Vδ2 T cell-dependent antileukemic response. It may, therefore, represent a potential candidate for the development of novel treatments for B-cell malignancies., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
10. MCLA-117, a CLEC12AxCD3 bispecific antibody targeting a leukaemic stem cell antigen, induces T cell-mediated AML blast lysis.
- Author
-
van Loo PF, Hangalapura BN, Thordardottir S, Gibbins JD, Veninga H, Hendriks LJA, Kramer A, Roovers RC, Leenders M, de Kruif J, Doornbos RP, Sirulnik A, Throsby M, Logtenberg T, Dolstra H, and Bakker ABH
- Subjects
- Animals, Antibodies, Bispecific metabolism, Antibodies, Bispecific pharmacokinetics, CD3 Complex immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cell Proliferation, Cytokines analysis, Cytokines metabolism, HL-60 Cells, Half-Life, Humans, Lectins, C-Type immunology, Leukemia, Myeloid, Acute immunology, Lymphocyte Activation, Mice, Mice, Inbred C57BL, Receptors, Mitogen immunology, T-Lymphocytes metabolism, Antibodies, Bispecific therapeutic use, Leukemia, Myeloid, Acute drug therapy, T-Lymphocytes immunology
- Abstract
Objective : We report the characterization of MCLA-117, a novel T cell-redirecting antibody for acute myeloid leukaemia (AML) treatment targeting CD3 on T cells and CLEC12A on leukaemic cells. In AML, CLEC12A is expressed on blasts and leukaemic stem cells. Methods : The functional capacity of MCLA-117 to redirect resting T cells to eradicate CLEC12A
POS tumor cells was studied using human samples, including primary AML samples. Results : Within the normal hematopoietic compartment, MCLA-117 binds to cells expressing CD3 and CLEC12A but not to early myeloid progenitors or hematopoietic stem cells. MCLA-117 induces T cell activation (EC50 = 44 ng/mL), T cell proliferation, mild pro-inflammatory cytokine release, and redirects T cells to lyse CLEC12APOS target cells (EC50 = 68 ng/mL). MCLA-117-induced targeting of normal CD34POS cells co-cultured with T cells spares erythrocyte and megakaryocyte differentiation as well as preserves mono-myelocytic lineage development. In primary AML patient samples with autologous T cells, MCLA-117 robustly induced AML blast killing (23-98%) at low effector-to-target ratios (1:3-1:97). Conclusion : These findings demonstrate that MCLA-117 efficiently redirects T cells to kill tumour cells while sparing the potential of the bone marrow to develop the full hematological compartment and support further clinical evaluation as a potentially potent treatment option for AML.- Published
- 2019
- Full Text
- View/download PDF
11. The architecture of EGFR's basal complexes reveals autoinhibition mechanisms in dimers and oligomers.
- Author
-
Zanetti-Domingues LC, Korovesis D, Needham SR, Tynan CJ, Sagawa S, Roberts SK, Kuzmanic A, Ortiz-Zapater E, Jain P, Roovers RC, Lajevardipour A, van Bergen En Henegouwen PMP, Santis G, Clayton AHA, Clarke DT, Gervasio FL, Shan Y, Shaw DE, Rolfe DJ, Parker PJ, and Martin-Fernandez ML
- Subjects
- Animals, CHO Cells, Cell Membrane metabolism, Cricetinae, Cricetulus, Extracellular Matrix metabolism, Fluorescent Dyes chemistry, Fluorescent Dyes metabolism, Ligands, Models, Biological, Models, Molecular, Photobleaching, Polymers chemistry, Protein Domains, Protein Kinases chemistry, Protein Kinases metabolism, ErbB Receptors antagonists & inhibitors, ErbB Receptors chemistry, Protein Multimerization
- Abstract
Our current understanding of epidermal growth factor receptor (EGFR) autoinhibition is based on X-ray structural data of monomer and dimer receptor fragments and does not explain how mutations achieve ligand-independent phosphorylation. Using a repertoire of imaging technologies and simulations we reveal an extracellular head-to-head interaction through which ligand-free receptor polymer chains of various lengths assemble. The architecture of the head-to-head interaction prevents kinase-mediated dimerisation. The latter, afforded by mutation or intracellular treatments, splits the autoinhibited head-to-head polymers to form stalk-to-stalk flexible non-extended dimers structurally coupled across the plasma membrane to active asymmetric tyrosine kinase dimers, and extended dimers coupled to inactive symmetric kinase dimers. Contrary to the previously proposed main autoinhibitory function of the inactive symmetric kinase dimer, our data suggest that only dysregulated species bear populations of symmetric and asymmetric kinase dimers that coexist in equilibrium at the plasma membrane under the modulation of the C-terminal domain.
- Published
- 2018
- Full Text
- View/download PDF
12. A bispecific nanobody approach to leverage the potent and widely applicable tumor cytolytic capacity of Vγ9Vδ2-T cells.
- Author
-
de Bruin RCG, Veluchamy JP, Lougheed SM, Schneiders FL, Lopez-Lastra S, Lameris R, Stam AG, Sebestyen Z, Kuball J, Molthoff CFM, Hooijberg E, Roovers RC, Santo JPD, van Bergen En Henegouwen PMP, Verheul HMW, de Gruijl TD, and van der Vliet HJ
- Abstract
Though Vγ9Vδ2-T cells constitute only a small fraction of the total T cell population in human peripheral blood, they play a vital role in tumor defense and are therefore of major interest to explore for cancer immunotherapy. Vγ9Vδ2-T cell-based cancer immunotherapeutic approaches developed so far have been generally well tolerated and were able to induce significant clinical responses. However, overall results were inconsistent, possibly due to the fact that these strategies induced systemic activation of Vγ9Vδ2-T cells without preferential accumulation and targeted activation in the tumor. Here we show that a novel bispecific nanobody-based construct targeting both Vγ9Vδ2-T cells and EGFR induced potent Vγ9Vδ2-T cell activation and subsequent tumor cell lysis both in vitro and in an in vivo mouse xenograft model. Tumor cell lysis was independent of KRAS and BRAF tumor mutation status and common Vγ9Vδ2-T cell receptor sequence variations. In combination with the conserved monomorphic nature of the Vγ9Vδ2-TCR and the facile replacement of the tumor-specific nanobody, this immunotherapeutic approach can be applied to a large group of cancer patients.
- Published
- 2017
- Full Text
- View/download PDF
13. High affinity nanobodies against human epidermal growth factor receptor selected on cells by E. coli display.
- Author
-
Salema V, Mañas C, Cerdán L, Piñero-Lambea C, Marín E, Roovers RC, Van Bergen En Henegouwen PM, and Fernández LÁ
- Subjects
- Animals, Escherichia coli, Humans, Peptide Library, Cell Surface Display Techniques methods, ErbB Receptors immunology, Single-Domain Antibodies isolation & purification
- Abstract
Most therapeutic antibodies (Abs) target cell surface proteins on tumor and immune cells. Cloning of Ab gene libraries in E. coli and their display on bacteriophages is commonly used to select novel therapeutic Abs binding target antigens, either purified or expressed on cells. However, the sticky nature of bacteriophages renders phage display selections on cells challenging. We previously reported an E. coli display system for expression of VHHs (i.e., nanobodies, Nbs) on the surface of bacteria and selection of high-affinity clones by magnetic cell sorting (MACS). Here, we demonstrate that E. coli display is also an attractive method for isolation of Nbs against cell surface antigens, such as the epidermal growth factor receptor (EGFR), upon direct selection and screening of Ab libraries on live cells. We employ a whole cell-based strategy using a VHH library obtained by immunization with human tumor cells over-expressing EGFR (i.e., A431), and selection of bacterial clones bound to murine fibroblast NIH-3T3 cells transfected with human EGFR, after depletion of non-specific clones on untransfected cells. This strategy resulted in the isolation of high-affinity Nbs binding distinct epitopes of EGFR, including Nbs competing with the ligand, EGF, as characterized by flow cytometry of bacteria displaying the Nbs and binding assays with purified Nbs using surface plasmon resonance. Hence, our study demonstrates that E. coli display of VHH libraries and selection on cells enables efficient isolation and characterization of high-affinity Nbs against cell surface antigens.
- Published
- 2016
- Full Text
- View/download PDF
14. Highly specific and potently activating Vγ9Vδ2-T cell specific nanobodies for diagnostic and therapeutic applications.
- Author
-
de Bruin RCG, Lougheed SM, van der Kruk L, Stam AG, Hooijberg E, Roovers RC, van Bergen En Henegouwen PMP, Verheul HMW, de Gruijl TD, and van der Vliet HJ
- Subjects
- Animals, Camelids, New World immunology, Cells, Cultured, Flow Cytometry methods, Humans, Immunohistochemistry methods, Immunomagnetic Separation methods, Jurkat Cells, Microscopy, Fluorescence, Receptors, Antigen, T-Cell, gamma-delta metabolism, Reproducibility of Results, T-Lymphocytes metabolism, Antibody Affinity immunology, Antibody Specificity immunology, Receptors, Antigen, T-Cell, gamma-delta immunology, Single-Domain Antibodies immunology, T-Lymphocytes immunology
- Abstract
Vγ9Vδ2-T cells constitute the predominant subset of γδ-T cells in human peripheral blood and have been shown to play an important role in antimicrobial and antitumor immune responses. Several efforts have been initiated to exploit these cells for cancer immunotherapy, e.g. by using phosphoantigens, adoptive cell transfer, and by a bispecific monoclonal antibody based approach. Here, we report the generation of a novel set of Vγ9Vδ2-T cell specific VHH (or nanobody). VHH have several advantages compared to conventional antibodies related to their small size, stability, ease of generating multispecific molecules and low immunogenicity. With high specificity and affinity, the anti-Vγ9Vδ2-T cell receptor VHHs are shown to be useful for FACS, MACS and immunocytochemistry. In addition, some VHH were found to specifically activate Vγ9Vδ2-T cells. Besides being of possible immunotherapeutic value, these single domain antibodies will be of great value in the further study of this important immune effector cell subset., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
15. Combining somatic mutations present in different in vivo affinity-matured antibodies isolated from immunized Lama glama yields ultra-potent antibody therapeutics.
- Author
-
Klarenbeek A, Blanchetot C, Schragel G, Sadi AS, Ongenae N, Hemrika W, Wijdenes J, Spinelli S, Desmyter A, Cambillau C, Hultberg A, Kretz-Rommel A, Dreier T, De Haard HJ, and Roovers RC
- Subjects
- Amino Acid Sequence, Animals, Antibody Affinity, Camelids, New World genetics, Humans, Immunoglobulin Fab Fragments chemistry, Interleukin-6 immunology, Models, Immunological, Models, Molecular, Recombinant Proteins chemistry, Sequence Alignment, Immunoglobulin Fab Fragments genetics, Immunoglobulin Fab Fragments metabolism, Mutation genetics, Peptide Library, Recombinant Proteins genetics, Recombinant Proteins metabolism
- Abstract
Highly potent human antibodies are required to therapeutically neutralize cytokines such as interleukin-6 (IL-6) that is involved in many inflammatory diseases and malignancies. Although a number of mutagenesis approaches exist to perform antibody affinity maturation, these may cause antibody instability and production issues. Thus, a robust and easy antibody affinity maturation strategy to increase antibody potency remains highly desirable. By immunizing llama, cloning the 'immune' antibody repertoire and using phage display, we selected a diverse set of IL-6 antagonistic Fabs. Heavy chain shuffling was performed on the Fab with lowest off-rate, resulting in a panel of variants with even lower off-rate. Structural analysis of the Fab:IL-6 complex suggests that the increased affinity was partly due to a serine to tyrosine switch in HCDR2. This translated into neutralizing capacity in an in vivo model of IL-6 induced SAA production. Finally, a novel Fab library was designed, encoding all variations found in the natural repertoire of VH genes identified after heavy chain shuffling. High stringency selections resulted in identification of a Fab with 250-fold increased potency when re-formatted into IgG1. Compared with a heavily engineered anti-IL-6 monoclonal antibody currently in clinical development, this IgG was at least equally potent, showing the engineering process to have had led to a highly potent anti-IL-6 antibody., (© The Author 2016. Published by Oxford University Press. All rights reserved. For Permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2016
- Full Text
- View/download PDF
16. Site-specific conjugation of single domain antibodies to liposomes enhances photosensitizer uptake and photodynamic therapy efficacy.
- Author
-
Broekgaarden M, van Vught R, Oliveira S, Roovers RC, van Bergen en Henegouwen PM, Pieters RJ, Van Gulik TM, Breukink E, and Heger M
- Subjects
- Animals, Cell Line, Tumor, Drug Carriers, Humans, Indoles chemistry, Isoindoles, Kinetics, Mice, Nanomedicine methods, Organometallic Compounds chemistry, Oxygen chemistry, Zinc chemistry, Zinc Compounds, Liposomes chemistry, Photochemotherapy methods, Photosensitizing Agents administration & dosage, Single-Domain Antibodies chemistry
- Abstract
Photodynamic therapy for therapy-resistant cancers will greatly benefit from targeted delivery of tumor photosensitizing agents. In this study, a strategy for the site-specific conjugation of single domain antibodies onto liposomes containing the photosensitizer zinc phthalocyanine was developed and tested.
- Published
- 2016
- Full Text
- View/download PDF
17. Camelid Ig V genes reveal significant human homology not seen in therapeutic target genes, providing for a powerful therapeutic antibody platform.
- Author
-
Klarenbeek A, El Mazouari K, Desmyter A, Blanchetot C, Hultberg A, de Jonge N, Roovers RC, Cambillau C, Spinelli S, Del-Favero J, Verrips T, de Haard HJ, and Achour I
- Subjects
- Animals, Camelids, New World, Camelus, Crystallography, X-Ray, Humans, Protein Structure, Tertiary, Immunoglobulin Variable Region chemistry, Immunoglobulin Variable Region genetics, Immunoglobulin Variable Region immunology, Protein Folding, Sequence Homology, Amino Acid
- Abstract
Camelid immunoglobulin variable (IGV) regions were found homologous to their human counterparts; however, the germline V repertoires of camelid heavy and light chains are still incomplete and their therapeutic potential is only beginning to be appreciated. We therefore leveraged the publicly available HTG and WGS databases of Lama pacos and Camelus ferus to retrieve the germline repertoire of V genes using human IGV genes as reference. In addition, we amplified IGKV and IGLV genes to uncover the V germline repertoire of Lama glama and sequenced BAC clones covering part of the Lama pacos IGK and IGL loci. Our in silico analysis showed that camelid counterparts of all human IGKV and IGLV families and most IGHV families could be identified, based on canonical structure and sequence homology. Interestingly, this sequence homology seemed largely restricted to the Ig V genes and was far less apparent in other genes: 6 therapeutically relevant target genes differed significantly from their human orthologs. This contributed to efficient immunization of llamas with the human proteins CD70, MET, interleukin (IL)-1β and IL-6, resulting in large panels of functional antibodies. The in silico predicted human-homologous canonical folds of camelid-derived antibodies were confirmed by X-ray crystallography solving the structure of 2 selected camelid anti-CD70 and anti-MET antibodies. These antibodies showed identical fold combinations as found in the corresponding human germline V families, yielding binding site structures closely similar to those occurring in human antibodies. In conclusion, our results indicate that active immunization of camelids can be a powerful therapeutic antibody platform.
- Published
- 2015
- Full Text
- View/download PDF
18. Targeting hepatocyte growth factor receptor (Met) positive tumor cells using internalizing nanobody-decorated albumin nanoparticles.
- Author
-
Heukers R, Altintas I, Raghoenath S, De Zan E, Pepermans R, Roovers RC, Haselberg R, Hennink WE, Schiffelers RM, Kok RJ, and van Bergen en Henegouwen PM
- Subjects
- Cell Line, Tumor, Female, Humans, Ovarian Neoplasms pathology, Proto-Oncogene Mas, Proto-Oncogene Proteins c-met immunology, Albumins metabolism, Endocytosis, Nanoparticles, Ovarian Neoplasms metabolism, Proto-Oncogene Proteins c-met metabolism, Single-Domain Antibodies immunology
- Abstract
The hepatocyte growth factor receptor (HGFR, c-Met or Met) is a receptor tyrosine kinase that is involved in embryogenesis, tissue regeneration and wound healing. Abnormal activation of this proto-oncogene product is implicated in the development, progression and metastasis of many cancers. Current therapies directed against Met, such as ligand- or, dimerization-blocking antibodies or kinase inhibitors, reduce tumor growth but hardly eradicate the tumor. In order to improve anti-Met therapy, we have designed a drug delivery system consisting of crosslinked albumin nanoparticles decorated with newly selected anti-Met nanobodies (anti-Met-NANAPs). The anti-Met NANAPs bound specifically to and were specifically taken up by Met-expressing cells and transported to lysosomes for degradation. Treatment of tumor cells with anti-Met NANAPs also resulted in downregulation of the total Met protein. This study shows that anti-Met NANAPs offer a potential system for lysosomal delivery of drugs into Met-positive tumor cells., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
19. Endocytosis of EGFR requires its kinase activity and N-terminal transmembrane dimerization motif.
- Author
-
Heukers R, Vermeulen JF, Fereidouni F, Bader AN, Voortman J, Roovers RC, Gerritsen HC, and van Bergen En Henegouwen PM
- Subjects
- Animals, Cell Line, Cell Membrane genetics, Dimerization, ErbB Receptors genetics, Humans, Mice, Phosphorylation, Protein Structure, Tertiary, Signal Transduction, Cell Membrane metabolism, Endocytosis, ErbB Receptors chemistry, ErbB Receptors metabolism
- Abstract
EGFR signaling is attenuated by endocytosis and degradation of receptor-ligand complexes in lysosomes. Endocytosis of EGFR is known to be regulated by multiple post-translational modifications. The observation that prevention of these modifications does not block endocytosis completely, suggests the involvement of other mechanism(s). Recently, receptor clustering has been suggested to induce internalization of multiple types of membrane receptors. However, the mechanism of clustering-induced internalization remains unknown. We have used biparatopic antibody fragments from llama (VHHs) to induce EGFR clustering without stimulating tyrosine kinase activity. Using this approach, we have found an essential role for the N-terminal GG4-like dimerization motif in the transmembrane domain (TMD) for clustering-induced internalization. Moreover, conventional EGF-induced receptor internalization depends exclusively on this TMD dimerization and kinase activity. Mutations in this dimerization motif eventually lead to reduced EGFR degradation and sustained signaling. We propose a novel role for the TMD dimerization motif in the negative-feedback control of EGFR. The widely conserved nature of GG4-like dimerization motifs in transmembrane proteins suggests a general role for these motifs in clustering-induced internalization.
- Published
- 2013
- Full Text
- View/download PDF
20. Structural evaluation of EGFR inhibition mechanisms for nanobodies/VHH domains.
- Author
-
Schmitz KR, Bagchi A, Roovers RC, van Bergen en Henegouwen PM, and Ferguson KM
- Subjects
- Antibodies, Monoclonal, Humanized chemistry, Antineoplastic Agents chemistry, Binding Sites, Binding, Competitive, Cetuximab, Crystallography, X-Ray, Cystine chemistry, ErbB Receptors antagonists & inhibitors, ErbB Receptors metabolism, Humans, Hydrogen Bonding, Hydrophobic and Hydrophilic Interactions, Models, Molecular, Protein Binding, Protein Structure, Tertiary, Single-Domain Antibodies metabolism, ErbB Receptors chemistry, Single-Domain Antibodies chemistry
- Abstract
The epidermal growth factor receptor (EGFR) is implicated in human cancers and is the target of several classes of therapeutic agents, including antibody-based drugs. Here, we describe X-ray crystal structures of the extracellular region of EGFR in complex with three inhibitory nanobodies, the variable domains of heavy chain only antibodies (VHH). VHH domains, the smallest natural antigen-binding modules, are readily engineered for diagnostic and therapeutic applications. All three VHH domains prevent ligand-induced EGFR activation, but use two distinct mechanisms. 7D12 sterically blocks ligand binding to EGFR in a manner similar to that of cetuximab. EgA1 and 9G8 bind an epitope near the EGFR domain II/III junction, preventing receptor conformational changes required for high-affinity ligand binding and dimerization. This epitope is accessible to the convex VHH paratope but inaccessible to the flatter paratope of monoclonal antibodies. Appreciating the modes of binding and inhibition of these VHH domains will aid in developing them for tumor imaging and/or cancer therapy., (Copyright © 2013 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
21. Amphiregulin enhances regulatory T cell-suppressive function via the epidermal growth factor receptor.
- Author
-
Zaiss DM, van Loosdregt J, Gorlani A, Bekker CP, Gröne A, Sibilia M, van Bergen en Henegouwen PM, Roovers RC, Coffer PJ, and Sijts AJ
- Subjects
- Amphiregulin, Animals, Antibodies, Neutralizing pharmacology, Cell Communication immunology, Colitis chemically induced, Colitis immunology, Colitis metabolism, Colitis pathology, EGF Family of Proteins, ErbB Receptors genetics, Forkhead Transcription Factors genetics, Forkhead Transcription Factors immunology, Gene Expression Regulation drug effects, Glycoproteins antagonists & inhibitors, Glycoproteins genetics, Glycoproteins pharmacology, Homeodomain Proteins genetics, Homeodomain Proteins immunology, Humans, Intercellular Signaling Peptides and Proteins genetics, Intercellular Signaling Peptides and Proteins pharmacology, Lymphocyte Activation drug effects, Mast Cells drug effects, Mast Cells immunology, Mast Cells metabolism, Melanoma, Experimental genetics, Melanoma, Experimental immunology, Melanoma, Experimental pathology, Membrane Proteins administration & dosage, Membrane Proteins immunology, Mice, Mice, Transgenic, Peptide Fragments administration & dosage, Peptide Fragments immunology, Signal Transduction drug effects, Signal Transduction immunology, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory metabolism, ErbB Receptors immunology, Glycoproteins immunology, Intercellular Signaling Peptides and Proteins immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Epidermal growth factor receptor (EGFR) is known to be critically involved in tissue development and homeostasis as well as in the pathogenesis of cancer. Here we showed that Foxp3(+) regulatory T (Treg) cells express EGFR under inflammatory conditions. Stimulation with the EGF-like growth factor Amphiregulin (AREG) markedly enhanced Treg cell function in vitro, and in a colitis and tumor vaccination model we showed that AREG was critical for efficient Treg cell function in vivo. In addition, mast cell-derived AREG fully restored optimal Treg cell function. These findings reveal EGFR as a component in the regulation of local immune responses and establish a link between mast cells and Treg cells. Targeting of this immune regulatory mechanism may contribute to the therapeutic successes of EGFR-targeting treatments in cancer patients., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
22. Therapeutic stem cells expressing variants of EGFR-specific nanobodies have antitumor effects.
- Author
-
van de Water JA, Bagci-Onder T, Agarwal AS, Wakimoto H, Roovers RC, Zhu Y, Kasmieh R, Bhere D, Van Bergen en Henegouwen PM, and Shah K
- Subjects
- Animals, Apoptosis immunology, Blotting, Western, Brain Neoplasms immunology, Brain Neoplasms pathology, Glioblastoma immunology, Glioblastoma pathology, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, HEK293 Cells, Humans, Immunoconjugates genetics, Immunoconjugates immunology, Immunoconjugates metabolism, Mice, Mice, Nude, Microscopy, Fluorescence, NIH 3T3 Cells, Neural Stem Cells immunology, Neural Stem Cells metabolism, Signal Transduction immunology, Single-Domain Antibodies genetics, Single-Domain Antibodies metabolism, TNF-Related Apoptosis-Inducing Ligand genetics, TNF-Related Apoptosis-Inducing Ligand immunology, TNF-Related Apoptosis-Inducing Ligand metabolism, Treatment Outcome, Tumor Burden immunology, Tumor Cells, Cultured, Brain Neoplasms therapy, ErbB Receptors immunology, Glioblastoma therapy, Neural Stem Cells transplantation, Single-Domain Antibodies immunology, Xenograft Model Antitumor Assays
- Abstract
The deregulation of the epidermal growth factor receptor (EGFR) has a significant role in the progression of tumors. Despite the development of a number of EGFR-targeting agents that can arrest tumor growth, their success in the clinic is limited in several tumor types, particularly in the highly malignant glioblastoma multiforme (GBM). In this study, we generated and characterized EGFR-specific nanobodies (ENb) and imageable and proapoptotic ENb immunoconjugates released from stem cells (SC) to ultimately develop a unique EGFR-targeted therapy for GBM. We show that ENbs released from SCs specifically localize to tumors, inhibit EGFR signaling resulting in reduced GBM growth and invasiveness in vitro and in vivo in both established and primary GBM cell lines. We also show that ENb primes GBM cells for proapoptotic tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis. Furthermore, SC-delivered immunoconjugates of ENb and TRAIL target a wide spectrum of GBM cell types with varying degrees of TRAIL resistance and significantly reduce GBM growth and invasion in both established and primary invasive GBM in mice. This study demonstrates the efficacy of SC-based EGFR targeted therapy in GBMs and provides a unique approach with clinical implications.
- Published
- 2012
- Full Text
- View/download PDF
23. Tumor-targeted Nanobullets: Anti-EGFR nanobody-liposomes loaded with anti-IGF-1R kinase inhibitor for cancer treatment.
- Author
-
van der Meel R, Oliveira S, Altintas I, Haselberg R, van der Veeken J, Roovers RC, van Bergen en Henegouwen PM, Storm G, Hennink WE, Schiffelers RM, and Kok RJ
- Subjects
- Animals, Antineoplastic Agents pharmacology, Binding, Competitive, Blotting, Western, Catechols pharmacology, Cell Line, Tumor, Cell Proliferation drug effects, Drug Compounding, Flow Cytometry, Humans, Immunoglobulin Heavy Chains pharmacology, Liposomes, Mice, Microscopy, Confocal, NIH 3T3 Cells, Nanoparticles, Particle Size, Protein Kinase Inhibitors pharmacology, Receptor Cross-Talk drug effects, Surface Properties, Tyrphostins pharmacology, Antineoplastic Agents administration & dosage, Catechols administration & dosage, ErbB Receptors antagonists & inhibitors, Immunoglobulin Heavy Chains administration & dosage, Protein Kinase Inhibitors administration & dosage, Receptor, IGF Type 1 antagonists & inhibitors, Tyrphostins administration & dosage
- Abstract
The epidermal growth factor receptor (EGFR) is a validated target for anti-cancer therapy and several EGFR inhibitors are used in the clinic. Over the years, an increasing number of studies have reported on the crosstalk between EGFR and other receptors that can contribute to accelerated cancer development or even acquisition of resistance to anti-EGFR therapies. Combined targeting of EGFR and insulin-like growth factor 1 receptor (IGF-1R) is a rational strategy to potentiate anti-cancer treatment and possibly retard resistance development. In the present study, we have pursued this by encapsulating the kinase inhibitor AG538 in anti-EGFR nanobody-liposomes. The thus developed dual-active nanobody-liposomes associated with EGFR-(over)expressing cells in an EGFR-specific manner and blocked both EGFR and IGF-1R activation, due to the presence of the EGFR-blocking nanobody EGa1 and the anti-IGF-1R kinase inhibitor AG538 respectively. AG538-loaded nanobody-liposomes induced a strong inhibition of tumor cell proliferation even upon short-term exposure followed by a drug-free wash-out period. Therefore, AG538-loaded nanobody-liposomes are a promising anti-cancer formulation due to efficient intracellular delivery of AG538 in combination with antagonistic and downregulating properties of the EGa1 nanobody-liposomes., (Copyright © 2011 Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
24. Rapid visualization of human tumor xenografts through optical imaging with a near-infrared fluorescent anti-epidermal growth factor receptor nanobody.
- Author
-
Oliveira S, van Dongen GA, Stigter-van Walsum M, Roovers RC, Stam JC, Mali W, van Diest PJ, and van Bergen en Henegouwen PM
- Subjects
- Animals, ErbB Receptors immunology, Female, Humans, Immunohistochemistry, Mice, Mice, Nude, NIH 3T3 Cells, Neoplasms immunology, ErbB Receptors metabolism, Neoplasms metabolism, Spectroscopy, Near-Infrared methods
- Abstract
Given that overexpression of the epidermal growth factor receptor (EGFR) is found in many types of human epithelial cancers, noninvasive molecular imaging of this receptor is of great interest. A number of studies have employed monoclonal antibodies as probes; however, their characteristic long half-life in the bloodstream has encouraged the development of smaller probes. In this study, an anti-EGFR nanobody-based probe was developed and tested in comparison with cetuximab for application in optical molecular imaging. To this aim, the anti-EGFR nanobody 7D12 and cetuximab were conjugated to the near-infrared fluorophore IRDye800CW. 7D12-IR allowed the visualization of tumors as early as 30 minutes postinjection, whereas with cetuximab-IR, no signal above background was observed at the tumor site. Quantification of the IR-conjugated proteins in the tumors revealed ≈ 17% of injected dose per gram 2 hours after injection of 7D12-IR, which was significantly higher than the tumor uptake obtained 24 hours after injection of cetuximab-IR. This difference is associated with the superior penetration and distribution of 7D12-IR within the tumor. These results demonstrate that this anti-EGFR nanobody conjugated to the NIR fluorophore has excellent properties for rapid preclinical optical imaging, which holds promise for its future use as a complementary diagnostic tool in humans.
- Published
- 2012
25. ErbB1 dimerization is promoted by domain co-confinement and stabilized by ligand binding.
- Author
-
Low-Nam ST, Lidke KA, Cutler PJ, Roovers RC, van Bergen en Henegouwen PM, Wilson BS, and Lidke DS
- Subjects
- Cell Line, Tumor, Dimerization, ErbB Receptors metabolism, Humans, Ligands, Protein Binding, ErbB Receptors chemistry, Protein Conformation
- Abstract
The extent to which ligand occupancy and dimerization contribute to erbB1 signaling is controversial. To examine this, we used two-color quantum-dot tracking for visualization of the homodimerization of human erbB1 and quantification of the dimer off-rate (k(off)) on living cells. Kinetic parameters were extracted using a three-state hidden Markov model to identify transition rates between free, co-confined and dimerized states. We report that dimers composed of two ligand-bound receptors are long-lived and their k(off) is independent of kinase activity. By comparison, unliganded dimers have a more than four times faster k(off). Transient co-confinement of receptors promotes repeated encounters and enhances dimer formation. Mobility decreases more than six times when ligand-bound receptors dimerize. Blockade of erbB1 kinase activity or disruption of actin networks results in faster diffusion of receptor dimers. These results implicate both signal propagation and the cortical cytoskeleton in reduced mobility of signaling-competent erbB1 dimers.
- Published
- 2011
- Full Text
- View/download PDF
26. A biparatopic anti-EGFR nanobody efficiently inhibits solid tumour growth.
- Author
-
Roovers RC, Vosjan MJ, Laeremans T, el Khoulati R, de Bruin RC, Ferguson KM, Verkleij AJ, van Dongen GA, and van Bergen en Henegouwen PM
- Subjects
- Animals, Antibodies, Monoclonal immunology, Antibodies, Monoclonal, Humanized, Antibody Affinity, Cell Line, Tumor, Cetuximab, Humans, Mice, Mice, Nude, Xenograft Model Antitumor Assays, Antibodies, Monoclonal pharmacology, Antibody Specificity, Carcinoma, Squamous Cell therapy, Epitopes, ErbB Receptors antagonists & inhibitors, ErbB Receptors immunology, Single-Chain Antibodies therapeutic use
- Abstract
The epidermal growth factor receptor (EGFR) has been shown to be a valid cancer target for antibody-based therapy. At present, several anti-EGFR monoclonal antibodies have been successfully used, such as cetuximab and matuzumab. X-ray crystallography data show that these antibodies bind to different epitopes on the ecto-domain of EGFR, providing a rationale for the combined use of these two antibody specificities. We have previously reported on the successful isolation of antagonistic anti-EGFR nanobodies. In our study, we aimed to improve the efficacy of these molecules by combining nanobodies with specificities similar to both cetuximab and matuzumab into a single biparatopic molecule. Carefully designed phage nanobody selections resulted in two sets of nanobodies that specifically blocked the binding of either matuzumab or cetuximab to EGFR and that did not compete for each others' binding. A combination of nanobodies from both epitope groups into the biparatopic nanobody CONAN-1 was shown to block EGFR activation more efficiently than monovalent or bivalent (monospecific) nanobodies. In addition, this biparatopic nanobody potently inhibited EGF-dependent cell proliferation. Importantly, in an in vivo model of athymic mice bearing A431 xenografts, CONAN-1 inhibited tumour outgrowth with an almost similar potency as the whole mAb cetuximab, despite the fact that CONAN-1 is devoid of an Fc portion that could mediate immune effector functions. Compared to therapy using bivalent, monospecific nanobodies, CONAN-1 was clearly more potent in tumour growth inhibition. These results show that the rational design of biparatopic nanobody-based anticancer therapeutics may yield potent lead molecules for further development., (Copyright © 2011 UICC.)
- Published
- 2011
- Full Text
- View/download PDF
27. Facile labelling of an anti-epidermal growth factor receptor Nanobody with 68Ga via a novel bifunctional desferal chelate for immuno-PET.
- Author
-
Vosjan MJ, Perk LR, Roovers RC, Visser GW, Stigter-van Walsum M, van Bergen En Henegouwen PM, and van Dongen GA
- Subjects
- Animals, Antibodies blood, Antibodies immunology, Buffers, Cell Line, Tumor, Chelating Agents chemistry, Cross-Linking Reagents chemistry, Deferoxamine chemistry, Drug Stability, Gallium Radioisotopes, Humans, Mice, Temperature, Antibodies chemistry, Deferoxamine analogs & derivatives, ErbB Receptors immunology, Isothiocyanates chemistry, Isotope Labeling methods, Positron-Emission Tomography methods, Radioimmunodetection methods
- Abstract
Purpose: The ∼15 kDa variable domains of camelid heavy-chain-only antibodies (called Nanobodies®) have the flexibility to be formatted as monovalent, monospecific, multivalent or multispecific single chain proteins with either fast or slow pharmacokinetics. We report the evaluation of the fast kinetic anti-epidermal growth factor receptor (EGFR) Nanobody 7D12, labelled with (68)Ga via the novel bifunctional chelate (BFC) p-isothiocyanatobenzyl-desferrioxamine (Df-Bz-NCS). Df-Bz-NCS has recently been introduced as the chelate of choice for (89)Zr immuno-positron emission tomography (PET)., Methods: Nanobody 7D12 was premodified with Df-Bz-NCS at pH 9. Radiolabelling with purified (68)Ga was performed at pH 5.0-6.5 for 5 min at room temperature. For in vitro stability measurements in storage buffer (0.25 M NaOAc with 5 mg ml(-1) gentisic acid, pH 5.5) at 4°C or in human serum at 37°C, a mixture of (67)Ga and (68)Ga was used. Biodistribution and immuno-PET studies of (68)Ga-Df-Bz-NCS-7D12 were performed in nude mice bearing A431 xenografts using (89)Zr-Df-Bz-NCS-7D12 as the reference conjugate., Results: The Df-Bz-NCS chelate was conjugated to Nanobody 7D12 with a chelate to Nanobody molar substitution ratio of 0.2:1. The overall (68)Ga radiochemical yield was 55-70% (not corrected for decay); specific activity was 100-500 MBq/mg. Radiochemical purity of the conjugate was >96%, while the integrity and immunoreactivity were preserved. (68/67)Ga-Df-Bz-NCS-7D12 was stable in storage buffer as well as in human serum during a 5-h incubation period (<2% radioactivity loss). In biodistribution studies the (68)Ga-labelled Nanobody 7D12 showed high uptake in A431 tumours (ranging from 6.1 ± 1.3 to 7.2 ± 1.5%ID/g at 1-3 h after injection) and high tumour to blood ratios, which increased from 8.2 to 14.4 and 25.7 at 1, 2 and 3 h after injection, respectively. High uptake was also observed in the kidneys. Biodistribution was similar to that of the reference conjugate (89)Zr-Df-Bz-NCS-7D12. Tumours were clearly visualized in a PET imaging study., Conclusion: Via a rapid procedure under mild conditions a (68)Ga-Nanobody was obtained that exhibited high tumour uptake and tumour to normal tissue ratios in nude mice bearing A431 xenografts. Fast kinetic (68)Ga-Nanobody conjugates can be promising tools for tumour detection and imaging of target expression.
- Published
- 2011
- Full Text
- View/download PDF
28. Downregulation of EGFR by a novel multivalent nanobody-liposome platform.
- Author
-
Oliveira S, Schiffelers RM, van der Veeken J, van der Meel R, Vongpromek R, van Bergen En Henegouwen PM, Storm G, and Roovers RC
- Subjects
- Animals, Carcinoma, Squamous Cell pathology, Carcinoma, Squamous Cell therapy, Cell Line, Tumor, Cell Proliferation drug effects, Epitopes genetics, Epitopes immunology, Epitopes metabolism, ErbB Receptors genetics, ErbB Receptors immunology, Female, Fluorescent Antibody Technique, Direct, Head and Neck Neoplasms pathology, Head and Neck Neoplasms therapy, Humans, Ligands, Liposomes immunology, Mice, Mice, Inbred BALB C, Mice, Nude, Protein Binding genetics, Protein Binding immunology, Tumor Burden drug effects, Xenograft Model Antitumor Assays methods, Down-Regulation drug effects, ErbB Receptors metabolism, Liposomes pharmacology
- Abstract
The epidermal growth factor receptor (EGFR) is a recognized target for tumor therapy and monoclonal antibodies (mAbs, e.g. cetuximab) have been developed to inhibit receptor activation. Besides blocking ligand (e.g. EGF) binding to the receptor, reports have shown that mAbs promote slow receptor internalization and degradation in lysosomes, i.e. downregulation. The efficacy of receptor downregulation was recently shown to depend on the size of receptor clusters formed at the cell surface. In this study, a multivalent platform is presented, consisting of nanobodies recognizing the ectodomain of EGFR (EGa1) coupled to PEG-liposomes, and the in vitro and in vivo effects of this system on EGFR internalization and downregulation were investigated. Nanobodies are the smallest functional antigen-binding immunoglobulin fragments and the EGa1 nanobody has been described as an EGFR-antagonist. EGa1-liposomes (EGa1-L) induced a more than 90% removal of EGFR from the cell surface, as a result of receptor internalization. Furthermore, this massive sequestration of EGFR mediated by EGa1-L lead to receptor degradation, while no degradation was detected with the monovalent nanobody. The downregulatory capacity here reported was found to be independent of the epitope on EGFR recognized by the grafted nanobody, and exclusive to the nanobody-liposomes, as anti-EGFR single chain variable fragments (scFv) coupled to liposomes were unable to induce this effect. Importantly, EGa1-L induced a significant inhibition of tumor cell proliferation, in vitro, an effect likely mediated by the combination of receptor downregulation and receptor antagonism. Also in vivo, EGFR downregulation was observed in tumors of mice intravenously injected with EGa1-L, indicating that this multivalent platform blocks ligand binding to the receptor and simultaneously induces the downregulation of EGFR., (Copyright (c) 2010 Elsevier B.V. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
29. Crosstalk between epidermal growth factor receptor- and insulin-like growth factor-1 receptor signaling: implications for cancer therapy.
- Author
-
van der Veeken J, Oliveira S, Schiffelers RM, Storm G, van Bergen En Henegouwen PM, and Roovers RC
- Subjects
- Antineoplastic Agents pharmacology, Drug Delivery Systems, Drug Discovery, Enzyme Activation, ErbB Receptors antagonists & inhibitors, Humans, Models, Biological, Receptor Cross-Talk drug effects, Receptor, IGF Type 1 antagonists & inhibitors, Signal Transduction drug effects, ErbB Receptors metabolism, Receptor Cross-Talk physiology, Receptor, IGF Type 1 metabolism, Signal Transduction physiology
- Abstract
Both the epidermal growth factor receptor (EGFR) and the insulin-like growth factor-1 receptor (IGF-1R) can contribute to tumor development and -progression through their effects on cell proliferation, inhibition of apoptosis, angiogenesis, anchorage-independent growth and tumor-associated inflammation. EGFR-targeting monoclonal antibodies and small molecule tyrosine kinase inhibitors are currently in clinical use for the treatment of several types of cancer. However, primary and acquired resistance to these agents often occurs and thereby limits the clinical efficacy of mono-specific targeted therapy. Results from both in vitro and in vivo studies indicate that cross-talk between EGFR and IGF-1R can lead to acquired resistance against EGFR-targeted drugs. This review describes the interface between the EGFR and IGF-1R signaling networks and the implications of the extensive cross-talk between these two receptor systems for cancer therapy. EGFR and IGF-1R interact on multiple levels, either through a direct association between the two receptors, by mediating the availability of each others ligands, or indirectly, via common interaction partners such as G protein coupled receptors (GPCR) or downstream signaling molecules. This multi-layered cross-talk and its involvement in the induction of resistance to targeted therapies provide a clear rationale for dual targeting of EGFR and IGF-1R. We discuss several (potential) strategies to simultaneously inhibit EGFR and IGF-1R signaling as promising novel therapeutic approaches.
- Published
- 2009
- Full Text
- View/download PDF
30. Selection and characterization of KDEL-specific VHH antibody fragments and their application in the study of ER resident protein expression.
- Author
-
Klooster R, Eman MR, le Duc Q, Verheesen P, Verrips CT, Roovers RC, and Post JA
- Subjects
- Amino Acid Sequence, Down-Regulation, HeLa Cells, Humans, Immunoglobulin Fragments isolation & purification, Immunoglobulin Heavy Chains isolation & purification, Immunoglobulin Variable Region isolation & purification, Molecular Sequence Data, Peptide Library, Protein Folding, Recombinant Proteins biosynthesis, Recombinant Proteins isolation & purification, Up-Regulation, Antibody Specificity, Endoplasmic Reticulum metabolism, Immunoglobulin Fragments biosynthesis, Immunoglobulin Heavy Chains biosynthesis, Immunoglobulin Variable Region biosynthesis, Receptors, Peptide immunology
- Abstract
Several diseases are caused by defects in the protein secretory pathway of the cell, particularly in the endoplasmic reticulum (ER). These defects are manifested by the activation of the unfolded protein response (UPR) that involves the transcriptional up-regulation of several ER resident proteins, the down-regulation of protein translation and up-regulation of ER associated degradation (ERAD). Although this transcriptional up-regulation of ER resident proteins during ER stress has been well described, data on differential protein expression of these same proteins are hardly available. Tools that would enable the simultaneous analysis of this set of proteins would be of high importance. Since the C-terminal KDEL sequence is a conserved epitope present in a large set of ER resident proteins, an antibody directed against this sequence would be such a tool. Using a carefully designed selection strategy, VHH antibody fragments from a non-immune phage display library were isolated that recognize the KDEL sequence at the C-terminus of proteins, irrespective of the protein context. In an accepted in vitro model for ER stress, this antibody was shown to be an excellent tool to study differences in ER resident protein expression. Furthermore, the application of this antibody showed differences in ER resident protein levels during replicative senescence of human umbilical vein endothelial cells (HUVECs), underlining its significance in biological research. The selection strategy used to obtain these KDEL-specific antibodies opens up ways to select antibodies to other conserved epitopes, such as the nuclear localization signal (NLS) or the peroxisomal targeting sequence, permitting the simultaneous analysis of specific groups of proteins.
- Published
- 2009
- Full Text
- View/download PDF
31. EGF induces coalescence of different lipid rafts.
- Author
-
Hofman EG, Ruonala MO, Bader AN, van den Heuvel D, Voortman J, Roovers RC, Verkleij AJ, Gerritsen HC, and van Bergen En Henegouwen PM
- Subjects
- Animals, ErbB Receptors analysis, Fluorescent Antibody Technique, G(M1) Ganglioside analysis, G(M1) Ganglioside metabolism, Glycosylphosphatidylinositols analysis, Glycosylphosphatidylinositols metabolism, Green Fluorescent Proteins analysis, Green Fluorescent Proteins metabolism, Humans, Mice, NIH 3T3 Cells, Signal Transduction, Epidermal Growth Factor pharmacology, ErbB Receptors metabolism, Membrane Microdomains metabolism
- Abstract
The suggestion that microdomains may function as signaling platforms arose from the presence of growth factor receptors, such as the EGFR, in biochemically isolated lipid raft fractions. To investigate the role of EGFR activation in the organization of lipid rafts we have performed FLIM analyses using putative lipid raft markers such as ganglioside GM1 and glycosylphosphatidylinositol (GPI)-anchored GFP (GPI-GFP). The EGFR was labeled using single domain antibodies from Llama glama that specifically bind the EGFR without stimulating its kinase activity. Our FLIM analyses demonstrate a cholesterol-independent colocalization of GM1 with EGFR, which was not observed for the transferrin receptor. By contrast, a cholesterol-dependent colocalization was observed for GM1 with GPI-GFP. In the resting state no colocalization was observed between EGFR and GPI-GFP, but stimulation of the cell with EGF resulted in the colocalization at the nanoscale level of EGFR and GPI-GFP. Moreover, EGF induced the enrichment of GPI-GFP in a detergent-free lipid raft fraction. Our results suggest that EGF induces the coalescence of the two types of GM1-containing microdomains that might lead to the formation of signaling platforms.
- Published
- 2008
- Full Text
- View/download PDF
32. Nanobodies in therapeutic applications.
- Author
-
Roovers RC, van Dongen GA, and van Bergen en Henegouwen PM
- Subjects
- Animals, Camelids, New World, Humans, Immunoglobulin Fragments chemistry, Immunoglobulin Fragments genetics, Immunoglobulin Heavy Chains chemistry, Immunoglobulin Heavy Chains genetics, Mice, Neoplasms immunology, Neoplasms therapy, Particle Size, Immunoglobulin Fragments therapeutic use, Immunoglobulin Heavy Chains therapeutic use, Protein Engineering
- Abstract
Over the years, many antibodies have been successfully generated to treat patients with life-threatening diseases, most notably cancer. While the first generation of antibodies, originating from mice, caused severe side effects and were relatively inefficient, technological advances have made it possible to obtain fully human antibodies for therapeutic use. 'Heavy-chain only' antibodies have recently been discovered in the blood of camelids. Because of their size, the antigen-binding units of these antibodies comprising only a single Ig fold are called Nanobodies. These antibody fragments have several remarkable features that make them ideal candidates as next-generation cancer therapeutics. Particularly appealing is their ability to simultaneously inhibit various crucial growth factor receptors or their ligands with a single molecule. In addition, they are easy to clone and express on the tip of filamentous phage, which opens the possibility to select for Nanobodies inducing particular biological effects. Nanobodies have potential to become important cancer therapeutics in the near future, displaying unequalled and unprecedented efficacies in treatment.
- Published
- 2007
33. Efficient inhibition of EGFR signaling and of tumour growth by antagonistic anti-EFGR Nanobodies.
- Author
-
Roovers RC, Laeremans T, Huang L, De Taeye S, Verkleij AJ, Revets H, de Haard HJ, and van Bergen en Henegouwen PM
- Subjects
- Animals, Antibodies isolation & purification, Antibody Formation, Antibody Specificity, Camelids, New World immunology, Cell Line, Tumor, Cell Proliferation drug effects, Dose-Response Relationship, Immunologic, Enzyme-Linked Immunosorbent Assay, ErbB Receptors immunology, Female, Humans, Immunoglobulin Heavy Chains isolation & purification, Ligands, Mice, Mice, Nude, Sensitivity and Specificity, Structure-Activity Relationship, Xenograft Model Antitumor Assays, Antibodies pharmacology, ErbB Receptors antagonists & inhibitors, Immunoglobulin Heavy Chains pharmacology, Signal Transduction drug effects
- Abstract
The development of a number of different solid tumours is associated with over-expression of ErbB1, or the epidermal growth factor receptor (EGFR), and this over-expression is often correlated with poor prognosis of patients. Therefore, this receptor tyrosine kinase is considered to be an attractive target for antibody-based therapy. Indeed, antibodies to the EGFR have already proven their value for the treatment of several solid tumours, especially in combination with chemotherapeutic treatment regimens. Variable domains of camelid heavy chain-only antibodies (called Nanobodies) have superior properties compared with classical antibodies in that they are small, very stable, easy to produce in large quantities and easy to re-format into multi-valent or multi-specific proteins. Furthermore, they can specifically be selected for a desired function by phage antibody display. In this report, we describe the successful selection and the characterisation of antagonistic anti-EGFR Nanobodies. By using a functional selection strategy, Nanobodies that specifically competed for EGF binding to the EGFR were isolated from "immune" phage Nanobody repertoires. The selected antibody fragments were found to efficiently inhibit EGF binding to the EGFR without acting as receptor agonists themselves. In addition, they blocked EGF-mediated signalling and EGF-induced cell proliferation. In an in vivo murine xenograft model, the Nanobodies were effective in delaying the outgrowth of A431-derived solid tumours. This is the first report describing the successful use of untagged Nanobodies for the in vivo treatment of solid tumours. The results show that functional phage antibody selection, coupled to the rational design of Nanobodies, permits the rapid development of novel anti-cancer antibody-based therapeutics.
- Published
- 2007
- Full Text
- View/download PDF
34. Ubiquilin recruits Eps15 into ubiquitin-rich cytoplasmic aggregates via a UIM-UBL interaction.
- Author
-
Regan-Klapisz E, Sorokina I, Voortman J, de Keizer P, Roovers RC, Verheesen P, Urbé S, Fallon L, Fon EA, Verkleij A, Benmerah A, and van Bergen en Henegouwen PM
- Subjects
- Adaptor Proteins, Signal Transducing, Animals, Autophagy-Related Proteins, COS Cells, Calcium-Binding Proteins chemistry, Calcium-Binding Proteins genetics, Carrier Proteins genetics, Cell Cycle Proteins genetics, Chlorocebus aethiops, Endosomal Sorting Complexes Required for Transport, HeLa Cells, Humans, Intracellular Signaling Peptides and Proteins, Phosphoproteins chemistry, Phosphoproteins genetics, Proteasome Endopeptidase Complex metabolism, Protein Structure, Tertiary, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Signal Transduction physiology, Amino Acid Motifs, Calcium-Binding Proteins metabolism, Carrier Proteins metabolism, Cell Cycle Proteins metabolism, Cytoplasm metabolism, Phosphoproteins metabolism, Ubiquitin metabolism
- Abstract
Eps15 and its related protein Eps15R are key components of the clathrin-mediated endocytic pathway. We searched for new binding partners of Eps15 using a yeast two-hybrid screen. We report here that ubiquilin (hPLIC1), a type-2 ubiquitin-like protein containing a ubiquitin-like domain (UBL) and a ubiquitin-associated domain (UBA), interacts with both Eps15 and Eps15R. Using glutathione-S-transferase pull-down experiments, we show that the first ubiquitin-interacting motif of Eps15 (UIM1) interacts directly with the UBL domain of ubiquilin, whereas it does not bind to ubiquitinated proteins. The second UIM of Eps15 (UIM2) binds poorly to the UBL domain but does bind to ubiquitinated proteins. Two other UIM-containing endocytic proteins, Hrs and Hbp, also interact with ubiquilin in a UIM-dependent manner, whereas epsin does not. Immunofluorescence analysis showed that endogenous Eps15 and Hrs, but not epsin, colocalize with green-fluorescent-protein-fused ubiquilin in cytoplasmic aggregates that are not endocytic compartments. We have characterized these green-fluorescent-protein-fused-ubiquilin aggregates as ubiquitin-rich intracytoplasmic inclusions that are recruited to aggresomes upon proteasome inhibition. Moreover, we show that endogenous Eps15 and endogenous ubiquilin colocalize to cytoplasmic aggregates and aggresomes. Finally, we show that the recruitment of Eps15 into ubiquilin-positive aggregates is UIM dependent. Altogether, our data identify ubiquilin as the first common UIM-binding partner of a subset of UIM-containing endocytic proteins. We propose that this UIM/UBL-based interaction is responsible for the sequestration of certain UIM-containing endocytic proteins into cytoplasmic ubiquitin-rich protein aggregates.
- Published
- 2005
- Full Text
- View/download PDF
35. Regulation of Tiam1-Rac signalling.
- Author
-
Mertens AE, Roovers RC, and Collard JG
- Subjects
- Animals, GTPase-Activating Proteins metabolism, Guanine Nucleotide Exchange Factors metabolism, Humans, Models, Biological, Protein Structure, Tertiary, Proteins chemistry, T-Lymphoma Invasion and Metastasis-inducing Protein 1, rho GTP-Binding Proteins metabolism, Proteins metabolism, Signal Transduction, rac GTP-Binding Proteins metabolism
- Abstract
The GTPases of the Rho family are molecular switches that play an important role in a wide range of cellular processes and are increasingly implicated in tumourigenesis. Unlike what was found for the Ras oncogenes in tumours, hardly any activating mutations have been found in the genes encoding Rho proteins. In the past, we have identified Tiam1 (T-lymphoma invasion and metastasis) as a specific activator for the Rho-like GTPase Rac. In vivo, Tiam1 deficiency protects against Ras-induced skin carcinogenesis, underscoring the consequences of deregulated signalling for the onset and progression of tumours. Thus, an important level of regulation of signalling via the Rho-like GTPases comes from the specific control of their activators. In this paper, we review what is known on the specific regulation of Tiam1 signalling towards Rac.
- Published
- 2003
- Full Text
- View/download PDF
36. Evidence for a bias toward intracellular antigens in the local humoral anti-tumor immune response of a colorectal cancer patient revealed by phage display.
- Author
-
Roovers RC, van der Linden E, Zijlema H, de Bruïne A, Arends JW, and Hoogenboom HR
- Subjects
- Enzyme-Linked Immunosorbent Assay, Escherichia coli metabolism, Flow Cytometry, Humans, Immunohistochemistry, Lymph Nodes immunology, Lymph Nodes pathology, Lymphocytes immunology, Peptide Library, Tetanus Toxoid metabolism, Tumor Cells, Cultured, Colorectal Neoplasms immunology
- Abstract
Many patients with colorectal carcinoma (CRC) mount a cellular as well as a humoral immune response to the tumor. To investigate the nature and specificity of the humoral immune response in a CRC patient, lymphocytes infiltrating the primary colorectal tumor and lymph nodes draining the tumor were used as antibody variable (V)-gene pools for the construction of phage antibody repertoires. These libraries were first validated by selection on the antigen tetanus toxoid and shown to contain antibodies that were probably derived from both naive and memory B cells. The repertoires were then screened for the presence of antibodies directed to CRC by selection on the cell line CaCo2. For comparison, the same selections were performed with a phage antibody repertoire made from B cells of healthy donors. Striking differences were observed in the panel of specificities selected from these different repertoires: although a large panel of antibodies reactive with patient-derived primary tumors was obtained from the immune repertoires, none of these discriminated between normal colonic epithelium and colon cancer and none were reactive with cell-surface antigens. However, selections using the non-immune library did result in numerous antibodies that recognized cell surface markers on CaCo2. These data suggest a bias in the local humoral immune response in this CRC patient, directed primarily toward intracellular epithelial-cell specific target antigens., (Copyright 2001 Wiley-Liss, Inc.)
- Published
- 2001
- Full Text
- View/download PDF
37. Identification of colon tumour-associated antigens by phage antibody selections on primary colorectal carcinoma.
- Author
-
Roovers RC, van der Linden E, de Bruïne AP, Arends JW, and Hoogenboom HR
- Subjects
- Biomarkers, Tumor analysis, Colorectal Neoplasms chemistry, DNA Fingerprinting, Escherichia coli immunology, Flow Cytometry, Humans, Tumor Cells, Cultured, Antigens, Neoplasm analysis, Colorectal Neoplasms diagnosis
- Abstract
Immunotargeting of solid tumours using antibodies has become a valuable tool for the detection of cancer metastases and the treatment of minimal residual disease. However, only few tumour antigens useful for targeting have been described to date. To identify cell-surface targets on colorectal carcinoma (CRC), we selected a large, human phage antibody repertoire on freshly isolated colon tumour cells. Two antibodies were identified that reacted with epithelial cell-restricted cell-surface antigens, whereas one clone preferentially reacted with stromal cells. These antigens are tumour-associated antigens, as shown by their uniform expression in tumours of different patients and of different differentiation stages and by their limited expression on normal tissues. The pattern of reactivity in immunohistochemistry (IHC) and enzyme-linked immunosorbent assay (ELISA) suggests that these antigens are different from previously identified tumour-associated antigens (e.g. Ep-CAM or c-ERB-2). This phage antibody-based method may lead to the cloning of novel tumour antigens that are useful for the immunotargeting of solid tumours.
- Published
- 2001
- Full Text
- View/download PDF
38. In vitro characterisation of a monovalent and bivalent form of a fully human anti Ep-CAM phage antibody.
- Author
-
Roovers RC, van der Linden E, de Bruïne AP, Arends JW, and Hoogenboom HR
- Subjects
- Amino Acid Sequence, Antigens, Neoplasm analysis, Antigens, Neoplasm chemistry, Base Sequence, Cell Adhesion Molecules analysis, Cell Adhesion Molecules chemistry, Colorectal Neoplasms chemistry, Cross Reactions, Epithelial Cell Adhesion Molecule, Humans, Immunoglobulin Fragments immunology, Immunotherapy, Molecular Sequence Data, Antibodies, Bispecific immunology, Antibodies, Neoplasm immunology, Antigens, Neoplasm immunology, Cell Adhesion Molecules immunology, Peptide Library
- Abstract
Antibodies to tumour-associated antigens are increasingly being used as targeting vehicles for the visualisation and for therapy of human solid tumours. The epithelial cell adhesion molecule (Ep-CAM) is an antigen that is overexpressed on a variety of human solid tumours and constitutes an attractive target for immunotargeting. We set out to obtain fully human antibodies to this antigen by selecting from a large antibody repertoire displayed on bacteriophages. Two single-chain variable antibody fragments (scFv) were identified that specifically bound recombinant antigen in vitro. One of the selected antibodies (VEL-1) cross-reacted with extracellular matrix components in immunohistochemistry of colon carcinoma, whereas the other scFv (VEL-2) specifically recognised colon cancer cells. The latter antibody was further characterised with respect to epitope specificity and kinetics of antigen-binding. It showed no competition with the well-characterised anti Ep-CAM MOC-31 monoclonal antibody and had an off-rate of 5 x 10(-2) s-1. To obtain an antibody format more suitable for in vivo tumour targeting and to increase the apparent affinity through avidity, the genes of scFv VEL-2 were re-formatted by fusion to a human (gamma 1) hinge region and CH3 domain. This "minibody" was expressed in Escherichia coli, specifically bound the Ep-CAM antigen and showed a 20-fold reduced off-rate in surface plasmon resonance analysis. These results show that phage antibody selection, combined with antibody engineering, may result in fully human antibody molecules with promising characteristics for in vivo use in tumour targeting.
- Published
- 2001
- Full Text
- View/download PDF
39. Design and application of diabodies, triabodies and tetrabodies for cancer targeting.
- Author
-
Todorovska A, Roovers RC, Dolezal O, Kortt AA, Hoogenboom HR, and Hudson PJ
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Bispecific chemistry, Antibodies, Bispecific genetics, Antibody Affinity, Dimerization, Humans, Immunoglobulin Fragments chemistry, Immunoglobulin Fragments genetics, Molecular Sequence Data, Antibodies, Bispecific therapeutic use, Immunoglobulin Fragments therapeutic use, Neoplasms therapy, Protein Engineering
- Abstract
Multivalent recombinant antibody fragments provide high binding avidity and unique specificity to a wide range of target antigens and haptens. This review describes the design and expression of diabodies, triabodies and tetrabodies using examples of scFv molecules that target viruses (influenza neuraminidase) and cancer (Ep-CAM; epithelial cell adhesion molecule). We discuss the preferred choice of linker length between V-domains to direct the formation of either diabodies (60 kDa), triabodies (90 kDa) or tetrabodies (120 kDa), each with size, flexibility and valency suited to different applications for in vivo imaging and therapy. The increased binding valency of these scFv multimers results in high avidity (low off-rates). A particular advantage for tumour targeting is that molecules of 60-100 kDa have increased tumour penetration and fast clearance rates compared to the parent Ig (150 kDa). We highlight a number of cancer-targeting scFv multimers that have recently successfully undergone pre-clinical trials for in vivo stability and efficacy. We also review the design of multi-specific Fv modules suited to cross-link two or more different target antigens. These bi- and tri-specific multimers can be formed by association of different scFv molecules and, in the first examples, have been designed as cross-linking reagents for T-cell recruitment into tumours (immunotherapy), viral retargeting (gene therapy) and as red blood cell agglutination reagents (immunodiagnostics).
- Published
- 2001
- Full Text
- View/download PDF
40. Human anti-CD30 recombinant antibodies by guided phage antibody selection using cell panning.
- Author
-
Klimka A, Matthey B, Roovers RC, Barth S, Arends JW, Engert A, and Hoogenboom HR
- Subjects
- Amino Acid Sequence, Animals, Antibody Affinity, Binding Sites, Antibody, Cloning, Molecular, Humans, Hybridomas, Immunoglobulin Variable Region genetics, Ki-1 Antigen biosynthesis, Ki-1 Antigen genetics, Mice, Molecular Sequence Data, Peptide Library, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Sequence Homology, Amino Acid, Immunoglobulin Variable Region immunology, Ki-1 Antigen immunology
- Abstract
In various clinical studies, Hodgkin's patients have been treated with anti-CD30 immunotherapeutic agents and have shown promising responses. One of the problems that appeared from these studies is the development of an immune response against the nonhuman therapeutics, which limits repeated administration and reduces efficacy. We have set out to make a recombinant, human anti-CD30 single-chain variable fragment (scFv) antibody, which may serve as a targeting moiety with reduced immunogenicity and more rapid tumour penetration in similar clinical applications. Rather than selecting a naive phage antibody library on recombinant CD30 antigen, we used guided selection of a murine antibody in combination with panning on the CD30-positive cell line L540. The murine monoclonal antibody Ki-4 was chosen as starting antibody, because it inhibits the shedding of the extracellular part of the CD30 antigen. This makes the antibody better suited for CD30-targeting than most other anti-CD30 antibodies. We have previously isolated the murine Ki-4 scFv by selecting a mini-library of hybridoma-derived phage scFv-antibodies via panning on L540 cells. Here, we report that phage display technology was successfully used to obtain a human Ki-4 scFv version by guided selection. The murine variable heavy (VH) and light (VL) chain genes of the Ki-4 scFv were sequentially replaced by human V gene repertoires, while retaining only the major determinant for epitope-specificity: the heavy-chain complementarity determining region 3 (CDR3) of murine Ki-4. After two rounds of chain shuffling and selection by panning on L540 cells, a fully human anti-CD30 scFv was selected. It competes with the parental monoclonal antibody Ki-4 for binding to CD30, inhibits the shedding of the extracellular part of the CD30 receptor from L540 cells and is thus a promising candidate for the generation of anti-CD30 immunotherapeutics.
- Published
- 2000
- Full Text
- View/download PDF
41. Guided selection of a pan carcinoma specific antibody reveals similar binding characteristics yet structural divergence between the original murine antibody and its human equivalent.
- Author
-
Beiboer SH, Reurs A, Roovers RC, Arends JW, Whitelegg NR, Rees AR, and Hoogenboom HR
- Subjects
- Amino Acid Sequence, Animals, Antibodies, Monoclonal biosynthesis, Antibodies, Monoclonal genetics, Antibody Affinity, Antigens, Neoplasm immunology, Base Sequence, Binding Sites, Antibody, Carcinoma pathology, Cloning, Molecular, Germ-Line Mutation genetics, Glycoproteins immunology, Humans, Immunoglobulin Variable Region biosynthesis, Immunoglobulin Variable Region chemistry, Immunoglobulin Variable Region genetics, Immunoglobulin Variable Region immunology, Immunoglobulin kappa-Chains biosynthesis, Immunoglobulin kappa-Chains chemistry, Immunoglobulin kappa-Chains genetics, Immunoglobulin kappa-Chains immunology, Kinetics, Mice, Models, Molecular, Molecular Sequence Data, Peptide Library, Recombinant Fusion Proteins biosynthesis, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins immunology, Antibodies, Monoclonal chemistry, Antibodies, Monoclonal immunology, Antibody Specificity immunology, Carcinoma immunology, Genetic Variation genetics, Protein Engineering methods
- Abstract
Antibody engineering provides an excellent tool for the generation of human immunotherapeutics for the targeted treatment of solid tumours. We have engineered and selected a completely human antibody to epithelial glycoprotein-2 (EGP-2), a transmembrane glycoprotein present on virtually all human simple epithelia and abundantly expressed on a variety of human carcinomas. We chose to use the procedure of "guided selection" to rebuild a high-affinity murine antibody into a human antibody, using two consecutive rounds of variable domain shuffling and phage library selection. As a starting antibody, the murine antibody MOC-31 was used. After the first round of guided selection, where the V(H) of MOC-31 was combined in Fab format with a human V(L)C(L) library, a small panel of human light chains was identified, originating from a segment of the VkappaIII family, whereas the MOC-31 V(L) is more homologous to the VkappaII family. Nevertheless, one of the chimaeric Fabs, C3, displayed an off-rate similar to MOC-31 scFv. Combining the V(L) of C3 with a human V(H) library, while retaining the V(H) CDR3 of MOC-31, clones were selected using human V(H) genes originating from the rarely used V(H)7 family. The best clone, 9E, shows over 13 amino acid mutations from the germline sequence, has an off-rate comparable to the original antibody and specifically binds to the "MOC-31"-epitope on EGP-2 in specificity and competition ELISA, FACS analysis and immunohistochemistry. In both V(L) and V(H) of antibody 9E, three germline mutations were found creating the MOC-31 homologue residue. Structural modelling of both murine and human antibodies reveals that one of the germline mutations, 53Y in V(H) CDR2, is likely to be involved in antigen binding. We conclude that, although they may bind the same epitope and have similar binding affinity to the antigen as the original murine antibody, human antibodies derived by guided selection unlike CDR-grafted antibodies, may retain only some of the original key elements of the binding site chemistry. The selected human anti-EGP-2 antibody will be a suitable reagent for tumour targeting., (Copyright 2000 Academic Press.)
- Published
- 2000
- Full Text
- View/download PDF
42. Model systems to study the parameters determining the success of phage antibody selections on complex antigens.
- Author
-
Mutuberria R, Hoogenboom HR, van der Linden E, de Bruïne AP, and Roovers RC
- Subjects
- Animals, Antigens immunology, Caco-2 Cells, Epithelial Cell Adhesion Molecule, Humans, Immunoglobulin Heavy Chains biosynthesis, Immunoglobulin Heavy Chains genetics, Immunoglobulin Heavy Chains immunology, Immunoglobulin Light Chains genetics, Immunoglobulin Light Chains immunology, Immunoglobulin Variable Region biosynthesis, Immunoglobulin Variable Region genetics, Immunoglobulin Variable Region immunology, Immunologic Techniques, Mice, Models, Immunological, Tumor Cells, Cultured, Antigens analysis, Antigens, Neoplasm immunology, Biomarkers, Tumor immunology, Cell Adhesion Molecules immunology, E-Selectin immunology, Peptide Library
- Abstract
Phage antibody display technology offers a powerful tool for the isolation of specific antibodies to defined target antigens. Most selection strategies described to date have relied on the availability of purified and often recombinant antigen, providing the possibility to perform selections on a well-defined antigen source. However, when the target antigen cannot be purified (e.g., an integral membrane protein), or if the antigen is unknown (e.g., when searching for novel markers on cells or tissues), panning of phage antibody libraries has to be performed on complex antigen sources such as cell surfaces or tissue sections, or even by in vivo selection methods. This provides a series of technical and experimental challenges. One focus of our research is to select antibodies directed to novel cancer-induced antigens expressed by tumours and by the tumour vasculature. To understand the parameters governing selection on complex antigen sources and to assess the efficiency of these phage library selections, we have set up two model selection systems in which both tumour cells and vascular endothelial cells serve as target "antigen". We describe a model based on phage antibodies directed to the tumour antigen epithelial glycoprotein-2, to compare phage antibody selections on a range of different antigen sources including purified and recombinant antigen, whole live cells, tissue cryosections and in vivo grown solid tumours. Secondly, we describe a model based on a phage antibody directed against the endothelial cell inducible adhesion molecule E-selectin. We compare selections on cultured cell monolayers with selections on cell suspensions immobilised on columns, to determine which selection approach is most suitable for the identification of novel tumour endothelial cell markers. Our data provide insight into the efficiency and thus potency of different selection strategies and show that there are very large differences in the recovery and enrichment of binding phage between the different methods tested. Our results further demonstrate the feasibility of phage antibody selections on whole, intact cells and show that these may sometimes compare favourably to selections on purified antigen. Selections on endothelial cells immobilised on columns compare favourably with selections on cell-monolayers; the most favourable conditions for both selection procedures are described. The implications of our data for phage antibody selections on these different complex antigen sources using either non-immune or immune phage antibody repertoires are discussed. The use of model systems such as the ones described here will help to determine optimal experimental conditions for phage library selections on complex antigens and aid in developing more powerful selection procedures for target discovery.
- Published
- 1999
- Full Text
- View/download PDF
43. A large non-immunized human Fab fragment phage library that permits rapid isolation and kinetic analysis of high affinity antibodies.
- Author
-
de Haard HJ, van Neer N, Reurs A, Hufton SE, Roovers RC, Henderikx P, de Bruïne AP, Arends JW, and Hoogenboom HR
- Subjects
- Amino Acid Sequence, Bacteriophages, Base Sequence, Biosensing Techniques, Cross Reactions, DNA Primers, Humans, Immunoglobulin Fab Fragments chemistry, Immunoglobulin Fragments chemistry, Immunoglobulin Fragments genetics, Immunoglobulin Variable Region genetics, Kinetics, Molecular Sequence Data, Restriction Mapping, Spleen chemistry, Antibodies isolation & purification, Immunoglobulin Fab Fragments genetics, Peptide Library
- Abstract
We report the design, construction, and use of the first very large non-immunized phage antibody library in Fab format, which allows the rapid isolation and affinity analysis of antigen-specific human antibody fragments. Individually cloned heavy and light chain variable region libraries were combined in an efficient two-step cloning procedure, permitting the cloning of a total of 3.7 x 10(10) independent Fab clones. The performance of the library was determined by the successful selection of on average 14 different Fabs against 6 antigens tested. These include tetanus toxoid, the hapten phenyl-oxazolone, the breast cancer-associated MUC1 antigen, and three highly related glycoprotein hormones: human chorionic gonadotropin, human luteinizing hormone, and human follicle-stimulating hormone. In the latter category, a panel of either homone-specific or cross-reactive antibodies were identified. The design of the library permits the monitoring of selections with polyclonal phage preparations and to carry out large scale screening of antibody off-rates with unpurified Fab fragments on BIAcore. Antibodies with off-rates in the order of 10(-2) to 10(-4) s-1 and affinities up to 2.7 nM were recovered. The kinetics of these phage antibodies are of the same order of magnitude as antibodies associated with a secondary immune response. This new phage antibody library is set to become a valuable source of antibodies to many different targets, and to play a vital role in target discovery and validation in the area of functional genomics.
- Published
- 1999
- Full Text
- View/download PDF
44. An anti-CD30 single-chain Fv selected by phage display and fused to Pseudomonas exotoxin A (Ki-4(scFv)-ETA') is a potent immunotoxin against a Hodgkin-derived cell line.
- Author
-
Klimka A, Barth S, Matthey B, Roovers RC, Lemke H, Hansen H, Arends JW, Diehl V, Hoogenboom HR, and Engert A
- Subjects
- Exotoxins genetics, Exotoxins pharmacology, Hodgkin Disease pathology, Humans, Hybridomas, Ki-1 Antigen therapeutic use, Peptide Fragments, Pseudomonas, Reed-Sternberg Cells drug effects, Tumor Cells, Cultured, Bacterial Toxins pharmacology, Hodgkin Disease immunology, Immunotoxins pharmacology, Ki-1 Antigen immunology, Recombinant Fusion Proteins genetics, Reed-Sternberg Cells immunology
- Abstract
The human CD30 receptor is highly overexpressed on the surface of Hodgkin Reed-Sternberg cells and has been shown to be an excellent target for selective immunotherapy using monoclonal antibody-based agents such as immunotoxins. To construct a new recombinant immunotoxin for possible clinical use in patients with Hodgkin's lymphoma, we have chosen the murine anti-CD30 hybridoma Ki-4 to generate a high-affinity Ki-4 single-chain variable fragment (scFv). Hybridoma V-genes were polymerase chain reaction-amplified, assembled, cloned and expressed as a mini-library for display on filamentous phage. Functional Ki-4 scFv were obtained by selection of binding phage on the Hodgkin lymphoma-derived, CD30-expressing cell line L540Cy. The selected recombinant Ki-4 scFv was shown to specifically bind to an overlapping epitope on the CD30 antigen with binding kinetics similar to those of the original antibody. The Ki-4 scFv was subsequently fused to a deletion mutant of Pseudomonas exotoxin A (ETA'). The resulting immunotoxin Ki-4(scFv)-ETA' specifically binds to CD30+ L540Cy cells and inhibits the protein synthesis by 50% at a concentration (IC50) of 43 pM. This recombinant immunotoxin is a promising candidate for further clinical evaluation in patients with Hodgkin's lymphoma or other CD30+ malignancies.
- Published
- 1999
- Full Text
- View/download PDF
45. Antibody phage display technology and its applications.
- Author
-
Hoogenboom HR, de Bruïne AP, Hufton SE, Hoet RM, Arends JW, and Roovers RC
- Subjects
- Animals, Bacteriophages ultrastructure, Cell Membrane chemistry, Genetic Techniques, Humans, Immunoglobulin Fragments genetics, Ligands, Antibodies, Viral genetics, Bacteriophages genetics, Biotechnology trends, Gene Library
- Abstract
In recent years, the use of display vectors and in vitro selection technologies has transformed the way in which we generate ligands, such as antibodies and peptides, for a given target. Using this technology, we are now able to design repertoires of ligands from scratch and use the power of phage selection to select those ligands having the desired (biological) properties. With phage display, tailor-made antibodies may be synthesized and selected to acquire the desired affinity of binding and specificity for in vitro and in vivo diagnosis, or for immunotherapy of human disease. This review addresses recent progress in the construction of, and selection from phage antibody libraries, together with novel approaches for screening phage antibodies. As the quality of large naïve and synthetic antibody repertoires improves and libraries becomes more generally available, new and exciting applications are pioneered such as the identification of novel antigens using differential selection and the generation of receptor a(nta)gonists. A combination of the design and generation of millions to billions of different ligands, together with phage display for the isolation of binding ligands and with functional assays for identifying (and possibly selecting) bio-active ligands, will open even more challenging applications of this inspiring technology, and provide a powerful tool for drug and target discovery well into the next decade.
- Published
- 1998
- Full Text
- View/download PDF
46. Construction and characterization of a bispecific diabody for retargeting T cells to human carcinomas.
- Author
-
Helfrich W, Kroesen BJ, Roovers RC, Westers L, Molema G, Hoogenboom HR, and de Leij L
- Subjects
- Animals, Antibodies, Bispecific metabolism, Antibody Specificity, Antigens, Neoplasm immunology, COS Cells metabolism, Chromatography, Gel, Electrophoresis, Polyacrylamide Gel, Humans, Immunization, Passive, Immunoglobulin Fragments metabolism, Lymphocyte Activation immunology, Neoplasms therapy, Recombinant Proteins biosynthesis, Recombinant Proteins metabolism, Recombinant Proteins pharmacology, Sodium Dodecyl Sulfate, T-Lymphocytes, Cytotoxic immunology, Antibodies, Bispecific biosynthesis, Antibodies, Bispecific pharmacology, Immunoglobulin Fragments biosynthesis, Immunoglobulin Fragments pharmacology, Neoplasms immunology, T-Lymphocytes immunology
- Abstract
We describe the construction of a recombinant bispecific antibody fragment in the diabody format with specificity for both the well-established human pancarcinoma associated target antigen EGP2 (epithelial glycoprotein 2, also known as the CO17-1A antigen or KSA) and the CD3epsilon chain of human TCR/CD3 complex. The murine anti-EGP2 (MOC31) single chain variable fragment (scFv) and the humanized anti-CD3 (Ucht1v9) scFv were cast into a diabody format (designated Dia5v9) using a short 5 amino acid Gly-Ser linker between immunoglobulin heavy-chain and light-chain variable domains. Purification of the poly-histidine tagged Dia5v9 was achieved from extracts of protease deficient Escherichia coli by IMAC chromatography. The Dia5v9 diabody showed strong binding to both EGP2 and CD3 in transfected cells. The in vitro efficacy of Dia5v9 in mediating tumor cell lysis by interleukin-2 activated human T cells appeared to be similar to that of the hybrid-hybridoma-derived BsF(ab')2 Bis1 (anti-EGP2/anti-CD3) in a standard 4-hr 51Cr-release assay. This small and partially humanized recombinant bispecific antibody fragment may be valuable for T-cell-based immunotherapeutical treatment protocols, retargeting activated peripheral blood T lymphocytes to lyse various human carcinomas in vivo.
- Published
- 1998
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.